Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Stem Cell Reports

Article

Activin Is Superior to BMP7 for Efficient Maintenance of Human iPSC-Derived


Nephron Progenitors
Shunsuke Tanigawa,1 Hidekazu Naganuma,1,2 Yusuke Kaku,1 Takumi Era,3 Tetsushi Sakuma,4
Takashi Yamamoto,4 Atsuhiro Taguchi,1,5 and Ryuichi Nishinakamura1,*
1Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
2Department of Urology, Kyushu University Graduate School of Medical Science, Fukuoka 812-8582, Japan
3Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
4Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima 739-8526, Japan
5Present address: Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany

*Correspondence: ryuichi@kumamoto-u.ac.jp
https://doi.org/10.1016/j.stemcr.2019.07.003

SUMMARY

Kidney formation is regulated by the balance between maintenance and differentiation of nephron progenitor cells (NPCs). Now that
directed differentiation of NPCs from human induced pluripotent stem cells (iPSCs) can be achieved, maintenance and propagation
of NPCs in vitro should be beneficial for regenerative medicine. Although WNT and FGF signals were previously shown to be essential
for NPC propagation, the requirement for BMP/TGFb signaling remains controversial. Here we reveal that activin has superior effects
to BMP7 on maintenance efficiency of human iPSC-derived NPCs. Activin expanded ITGA8+/PDGFRA/SIX2-GFP+ NPCs by 5-fold per
week at 80%–90% efficiency, and the propagated cells possessed robust capacity for nephron formation both in vitro and in vivo. The
expanded cells also maintained their nephron-forming potential after freezing. Furthermore, the protocol was applicable to multiple
non-GFP-tagged iPSC lines. Thus, our activin-based protocol will be applicable to a variety of research fields including disease modeling
and drug screening.

INTRODUCTION these goals. Indeed, several groups, including ours, have


reported expansion protocols for NPCs derived from
The kidney contains a large number of nephrons, which ESCs or iPSCs (Brown et al., 2015; Li et al., 2016; Tanigawa
are functional units consisting of glomeruli and renal tu- et al., 2015, 2016), all of which involved fibroblast growth
bules. The kidney develops by interactions between the factor (FGF) and WNT signals, consistent with previous
metanephric mesenchyme and the ureteric bud, the former findings in vivo (Barak et al., 2012). However, the utiliza-
of which contains nephron progenitor cells (NPCs) that tion of bone morphogenetic protein (BMP) signaling mol-
form the epithelia of glomeruli and renal tubules in ecules and their inhibitors differed depending on the
response to wingless-type mouse mammary tumor virus protocol. Brown et al. (2015) reported expansion of hu-
integration site family member (WNT) signals emitted man iPSC-derived NPCs by addition of BMP7 and BMP re-
from the latter. At the same time, the ureteric bud-derived ceptor inhibitor LDN193189. However, their NPCs only
signals maintain and propagate some NPCs in the undiffer- formed renal tubule-like structures and not glomeruli.
entiated state, and the balance between NPC maintenance/ When the same protocol was applied to NPCs from hu-
propagation and differentiation is critical for kidney devel- man embryos, some NPC genes were maintained, but
opment (Costantini and Kopan, 2010). NPCs cease propa- the expanded cells failed to show nephron formation his-
gation and become terminally differentiated within a few tologically (Pode-Shakked et al., 2017). We reported
days after birth in mice (Hartman et al., 2007) and at expansion of NPCs from mouse embryos by adding leuke-
approximately 36 weeks of gestation in humans (Lind- mia inhibitory factor (LIF) and low concentrations of
strom et al., 2018a). BMP7. This protocol was applicable to NPCs derived
In recent years, we and others have succeeded in NPC from mouse ESCs and human iPSCs, and generated both
induction and resultant kidney organoid generation glomeruli and renal tubules, emphasizing that the full po-
from mouse embryonic stem cells (ESCs) and/or human tency for nephron formation was maintained in the
induced pluripotent stem cells (iPSCs) (Morizane et al., expanded NPCs. However, the expansion period for hu-
2015; Taguchi et al., 2014; Taguchi and Nishinakamura, man iPSC-derived NPCs was limited (1–2 weeks). Subse-
2017; Takasato et al., 2016). Thus, NPC purification and quently, Li et al. (2016) reported long-term expansion of
expansion should have an impact on regenerative NPCs from mouse and human embryos. They utilized a
medicine targeting cell therapy, disease modeling, drug spheroid culture in the presence of LIF, BMP7, and combi-
screening, and transplantable kidney reconstruction, nation of BMP and transforming growth factor b (TGFb)
because large numbers of NPCs are required to achieve receptor inhibitors, and obtained propagation of human

322 Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019 j ª 2019 The Author(s).
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Figure 1. NPCs Are Enriched in the ITGA8+/PDGFRA/SIX2-GFP+ Fraction in Human iPSC-Derived Kidney Tissues
(A) Generation of SIX2-GFP iPSCs by TALENs. A GFP cassette was inserted into exon 1 in the SIX2 gene by homologous recombination. The
selection cassette was removed by Cre-mediated excision. Southern blot analysis of wild-type (WT) (+/+), heterozygous (+/), and
homozygous (/) clones (right). The positions of the probe for Southern blotting and PCR primers are indicated on the map (left).
(B) Isolation of nephron progenitors from the ITGA8+/PDGFRA/SIX2-GFP+ fraction in induced NPCs. The gates in the dot plots show SIX2-
GFP+ cells in the total cells (left), ITGA8+/PDGFRA cells in the total cells (center), and ITGA8+/SIX2-GFP+ cells in the ITGA8+/PDGFRA
fraction (right). I, ITGA8; P, PDGFRA; SIX2+, SIX2-GFP+. The percentage of ITGA8+/PDGFRA/SIX2-GFP+ cells in the total cells is shown on
the right (I+/P/SIX2+ cells/total cells). Data represent means ± SEM from six independent experiments.
(legend continued on next page)

Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019 323


embryo-derived NPCs for 7 months. They further claimed structed a pair of plasmids expressing TALENs that tar-
that the same protocol was applicable to human iPSC- geted regions in close proximity to the SIX2 start codon,
derived NPCs. However, none of these reports addressed and introduced these plasmids together with a targeting
the quantitative percentages of human NPCs. If NPCs vector containing the GFP gene and homology arms into
constitute only a minor population of the expanded cells, iPSCs (Figure 1A). We successfully obtained heterozygous
nephrogenesis will occur in limited areas of the tissues, GFP-knockin clones and induced them toward NPCs,
and thus hamper reproducible disease modeling or drug based on our previously published protocol (Taguchi
screening. Thus, it is necessary to establish methods that et al., 2014; Taguchi and Nishinakamura, 2017). At day
can measure the precise percentages of functional NPCs 13, SIX2-GFP+ cells constituted 7.2% of the induced
in expanded cells. tissues (n = 6; Figure 1B). Meanwhile, the ITGA8+/
The transcription factor SIX2 is specifically expressed in PDGFRA population represented 22.7%, and 18.1%
NPCs and antagonizes their Wnt-mediated differentiation of these cells were SIX2-GFP+. Thus, 4.1% of the total
(Kobayashi et al., 2008). Li et al. (2016) generated SIX2- cells were ITGA8+/PDGFRA/SIX2-GFP+. Expression of
GFP iPSCs, but did not perform quantitative analysis of NPC marker Eya1 in ITGA8+/PDGFRA/SIX2-GFP+ or
GFP+ NPCs. Meanwhile, we reported that NPCs from ITGA8+/PDGFRA/SIX2-GFP cells was higher than that
mouse embryos and human iPSCs could be sorted as an in ITGA8/PDGFRA/SIX2-GFP cells, while the expres-
ITGA8+/PDGFRA population (Kaku et al., 2017; Taguchi sion levels of other NPC markers, such as SIX2,
et al., 2014). ITGA8 was expressed in NPCs from both MEOX1, and HOXA11, were higher in ITGA8+/
mice and humans in vivo (O’Brien et al., 2016), and deletion PDGFRA/SIX2-GFP+ cells even when compared with
of Itga8, as well as its ligand nephronectin, led to kidney hy- ITGA8+/PDGFRA/SIX2-GFP cells (Figure 1C). When
poplasia in mice (Muller et al., 1997), indicating that ITGA8 these fractions were aggregated and combined with em-
expression in NPCs is critical for kidney development. Sin- bryonic spinal cord, a classical inducer of nephrogenesis,
gle-cell RNA-sequencing (RNA-seq) analysis of nephron ITGA8+/PDGFRA/SIX2-GFP+ cells formed nephron
progenitors in human embryos further revealed that they epithelia of glomeruli and renal tubules (CADHERIN6
were ITGA8+/PDGFRA (Lindstrom et al., 2018b; O’Brien [CDH6]: proximal tubules; NEPHRIN: podocytes;
et al., 2016). Therefore, we reasoned that the combination E-CADHERIN [CDH1]: distal tubules) that constituted
of these two criteria (SIX2-GFP+ and ITGA8+/PDGFRA) 53.3% of the induced kidney tissues (Figures 1D and
would lead to more reliable and quantitative readouts for 1E). In contrast, ITGA8+/PDGFRA/SIX2-GFP cells ex-
measurement of maintenance efficiency of NPCs upon cul- hibited minimal nephrogenesis (8.7% of the whole tis-
ture. For this purpose, we generated SIX2-GFP human sues). The ITGA8/PDGFRA/SIX2-GFP fraction also
iPSCs and measured the percentages of ITGA8+/ showed minimal nephrogenesis (Figure S1), consistent
PDGFRA/SIX2-GFP+ NPCs, leading to the unexpected with our previous report (Kaku et al., 2017). Thus, hu-
finding that activin A (activin) has superior effects to man iPSC-derived NPCs were preferentially populated
BMP7 on the maintenance efficiency of human iPSC- in the ITGA8+/PDGFRA/SIX2-GFP+ fraction.
derived NPCs.

BMP7 Is Insufficient to Maintain Human iPSC-Derived


RESULTS NPCs
To propagate iPSC-derived ITGA8+/PDGFRA/SIX2-GFP+
Human iPSC-Derived NPCs Are Enriched in the cells, we aggregated the sorted cells and cultured them as
ITGA8+/PDGFRA/SIX2-GFP+ Fraction spheres for 7 days under our previously defined conditions,
To isolate NPCs, we inserted GFP into the SIX2 locus of which included FGF9, CHIR99021 (CHIR), LIF, DAPT
human iPSCs by homologous recombination. We con- (N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine

(C) Progenitor marker expression in each progenitor fraction. Isolated cells from the indicated fractions were analyzed for gene expression
by qPCR. Data represent means ± SEM from three independent experiments. *p < 0.05, **p < 0.01 versus ITGA8/PDGFRA/SIX2-GFP; NS,
not significant.
(D) Histology and immunostaining of spinal cord-induced tissues from isolated NPCs at day 9 after induction. Magnified images of the
ITGA8+/PDGFRA/SIX2-GFP+ fraction are shown on the right. CDH6 (green) shows proximal tubules, NEPHRIN (red) shows glomeruli, and
CDH1 (yellow) shows distal tubules. Scale bars: 200 mm (left and middle) and 50 mm (right).
(E) Quantification of nephron areas in induced tissues. The percentages of nephron structures in the induced tissues relative to the total
section areas were calculated and shown as bar graphs. **p < 0.01 versus ITGA8+/PDGFRA/SIX2-GFP (n = 3 per group).
See also Figure S1.

324 Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019


Figure 2. BMP7 Is Insufficient to Maintain
Human iPSC-Derived NPCs
(A) Schematic diagram of the culture pro-
cedure for purified NPCs. Purified NPCs were
aggregated and cultured in basal medium
containing FGF9, CHIR, LIF, DAPT, and BMP7
(5 or 50 ng/mL) for 7 days.
(B) Flow-cytometry quantification of
ITGA8+/PDGFRA/SIX2-GFP+ (I+/P/SIX2+)
cells after culture with BMP7 (5 ng/mL) for
7 days. The percentage of ITGA8+/PDGFRA/
SIX2-GFP+ cells in the total cells is shown on
the right (I+/P/SIX2+ cells/total cells).
n = 3 per group.
(C) Flow-cytometry quantification of ITGA8+/
PDGFRA/SIX2-GFP+ cells after culture with
BMP7 (50 ng/mL) for 7 days (n = 3 per
group).
(D) Numbers of total and ITGA8+/PDGFRA/
SIX2-GFP+ cells after 7 days of culture (n = 3
per group).
(E) qPCR analysis of progenitor markers in
NPCs after culture for 7 days. Freshly isolated
ITGA8+/PDGFRA/SIX2-GFP+ cells were used
as a positive control (Day 0). Data represent
means ± SEM from three independent ex-
periments. *p < 0.05, **p < 0.01, cells
cultured with BMP7 versus Day 0.
See also Figure S2.

t-butyl ester), and BMP7 (Figure 2A) (Tanigawa et al., 2016). 24.1% and 26.9% of the total cells, respectively (Figures
We previously proposed that a relatively low concentration 2B and 2C). Although the cell numbers increased signifi-
of BMP7 (5 ng/mL) was beneficial for maintenance cantly during the 7-day culture, the expansion of the
of NPCs, while other reports utilized higher concentrations ITGA8+/SIX2-GFP+ fraction was only 2.7-fold and 1.9-
(30–50 ng/mL) along with BMP receptor inhibitors (Barak fold, respectively (Figure 2D). Consistent with these obser-
et al., 2012; Brown et al., 2015; Li et al., 2016). Thus, vations, the expression levels of NPC markers in the whole
we examined the effects of two concentrations of BMP7 spheres decreased while those of differentiation markers
(5 ng/mL versus 50 ng/mL). However, the percentages (LHX1, CDH1) increased (Figure 2E). Thus, BMP7-based
of SIX2-GFP+ cells were 70.7% and 58.3%, respectively, conditions were insufficient for maintenance of ITGA8+/
and ITGA8+/PDGFRA/SIX2-GFP+ cells constituted only PDGFRA/SIX2-GFP+ NPCs in vitro, although the lower

Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019 325


Figure 3. Activin Maintains NPCs at High Efficiency
(A) Quantification of ITGA8+/PDGFRA/SIX2-GFP+ (I+/P/SIX2+) cells cultured for 7 days with BMP7 or TGFb-related ligands. NPCs were
cultured for 7 days in basal medium (as shown in Figure 2A) with addition of BMP7 (50 ng/mL) or TGFb-related ligands activin (50 ng/mL),
GDF11 (50 ng/mL), TGFb1 (50 ng/mL), or TGFb2 (50 ng/mL), then analyzed by flow cytometry. The percentage of ITGA8+/PDGFRA/SIX2-
GFP+ cells in the total cells is shown on the right (I+/P/SIX2+ cells/total cells). n = 3 per group.
(B) Numbers of total and ITGA8+/PDGFRA/SIX2-GFP+ cells after culture for 7 days (n = 3 per group).
(C) qPCR analysis of NPC markers. Freshly isolated ITGA8+/PDGFRA/SIX2-GFP+ cells were used as a positive control (Day 0). Data represent
means ± SEM from three independent experiments. **p < 0.01 versus day 0.
(D) Reconstitution of three-dimensional nephron structures by NPCs cultured with BMP7 or activin for 7 days. H&E-stained images (upper)
and immunostained images (lower) of induced nephrons from NPCs cultured with BMP7 or activin are shown. Scale bars, 200 mm.
(E) Quantification of nephron areas in induced tissues. The percentages of nephron-like structures in the induced tissues related the total
section areas were calculated and shown as bar graphs. Data represent means ± SEM from three independent experiments. **p < 0.01, cells
cultured with BMP7 versus activin.
(F) Magnified images of glomerular podocytes generated from NPCs expanded with activin. COL4, type IV collagen. Arrowheads indicate
pre-slit diaphragm domains; arrows indicate basal pre-slit diaphragm domains. Scale bar, 10 mm.
(G) H&E staining of kidney tissues generated from thawed NPCs at day 20 after spinal cord induction. Scale bar, 200 mm.

(legend continued on next page)

326 Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019


BMP7 concentration showed slightly better degrees of tion markers (LHX1, CDH1, HNF1B, JAG1, GATA3, and
propagation and NPC marker retention. EMX2) were upregulated. Therefore, activin can maintain
Li et al. (2016) reported that the combination of BMP7, human iPSC-derived ITGA8+/PDGFRA/SIX2-GFP+ NPCs
BMP receptor inhibitor LDN193189, and TGFb receptor in- more effectively than BMP7. Next, we examined the
hibitor A83-01 was effective for 7-month in vitro expansion nephron-forming potential of the expanded cells. Cells
of NPCs derived from human embryos. When we applied expanded with activin or other TGFb-related ligands ex-
their conditions to our iPSC-derived ITGA8+/PDGFRA/ hibited robust formation of glomeruli and renal tubules
SIX2-GFP+ cells, robust cell proliferation was observed at day 20 after spinal cord induction, and the areas of
and 71.5% of the cells remained SIX2-GFP+ (Figures S2A nephrogenesis were much larger than that for cells treated
and S2B). However, the percentages of the ITGA8+/ with BMP7 (Figures 3D and 3E, n = 6; Figure S4). Among
PDGFRA fraction were low, even at day 7 of culture them, activin showed the largest area of nephrogenesis
(25.5%), and only 18.2% of the expanded cells remained (Figure S4, n = 3), and was thus selected for use in subse-
ITGA8+/PDGFRA/SIX2-GFP+. RT-PCR analysis showed quent experiments. Notably, NEPHRIN+ rod or dot-like
that SIX2 expression was relatively maintained, consistent structures were observed in the glomerular podocytes
with the high SIX2-GFP+ cell percentage, but Eya1 was generated from activin-expanded NPCs. These structures
reduced and a differentiation marker, CDH1, was increased were detected on the lateral domains of the podocytes,
at day 7 (Figure S2C). Therefore, monitoring of SIX2 expres- as well as on their basal domains adjacent to the type IV
sion alone could lead to over-estimation of the mainte- collagen (COL4)-positive basement membrane (Figure 3F).
nance efficiency of NPCs. We recently reported that these structures observed in
iPSC-derived kidney organoids represent the precursors
Activin Maintains NPCs at High Efficiency of slit diaphragms, which play a critical role in filtration
Among the various reagents tested for improvement of the of blood to produce urine (Tanigawa et al., 2018). Thus,
culture condition, such as BMP4, FGF10, FGF20, CXCL12, the activin-based protocol can retain NPCs at comparable
Wnt9b, and R-spondin, we found that TGFb-related li- quality to the start of the culture for the generation of
gands, including activin, GDF11, TGFb1, and TGFb2, led these nephron structures.
to better maintenance of ITGA8+/PDGFRA/SIX2-GFP+ We further examined whether the expanded NPCs could
cells than BMP7 at day 7 of culture (Figure 3A). Among be frozen before induction to form nephrons. NPCs were
them, activin was the most effective: 91.6% of cells were cultured for 7 days with activin, frozen in single-cell sus-
SIX2-GFP+ and up to 83.1% of cells remained ITGA8+/ pensions, and thawed for regeneration of NPC spheres.
PDGFRA/SIX2-GFP+ (Figures 3A and S3). Furthermore, ac- When the spheres were induced with spinal cord they
tivin achieved 5.3 ± 0.8-fold expansion (n = 3) of NPCs, readily exhibited nephrogenesis, including glomerular po-
which was superior to BMP7 (Figure 3B). The expression docytes with slit diaphragm precursors (Figures 3G–3I). The
levels of SIX2 and other progenitor markers, as well as dif- nephron areas constituted 79.8% ± 3.6% (n = 4), and were
ferentiation markers, were comparable with those at the comparable with those of unfrozen progenitors. These re-
starting point (day 0), in sharp contrast to those with sults suggest that the complicated iPSC handling required
BMP7 treatment (Figure 3C). Comprehensive RNA-seq for NPC induction can be bypassed by starting with frozen
and clustering analyses also placed the NPCs cultured NPC stocks.
with activin closer to those at the start of culture and in
human embryos, compared with the NPCs cultured with Each of the Added Factors Is Required for Proper NPC
BMP7 (Figure 4). The activin-based protocol maintained Expansion
nephron progenitor markers, including SIX2, SALL1, To confirm the requirement of each added factor for NPC
EYA1, CITED1, PAX2, WT1, MEOX1, and ITGA8. Human maintenance and propagation, we excluded each factor
NPC-specific genes, such as LYPD1, TMEM100, WASF3, from the optimized medium and performed assessment
UNC5B, and PHF19, which were recently identified in hu- on day 7 of culture (Figure 5A). When activin was elimi-
man embryos (Lindstrom et al., 2018b), were also main- nated, the percentage of the ITGA8+/PDGFRA/SIX2-
tained. In contrast, SIX2 and MEOX1 were significantly GFP+ fraction decreased dramatically to 42% as expected,
downregulated upon BMP7 treatment while differentia- while the non-progenitor cell numbers were increased

(H) Immunostaining of nephrons generated from thawed NPCs. CDH6 (green), NEPHRIN (red), and CDH1 (yellow) are shown. Scale bar,
200 mm.
(I) Magnified image of glomerular podocytes. Immunostaining for NEPHRIN (red) and COL4 (green) are shown. Arrowheads indicate pre-
slit diaphragm domains; arrows indicate basal pre-slit diaphragm domains. Scale bar, 10 mm.
See also Figures S3 and S4.

Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019 327


Figure 4. RNA-Seq Analysis of the Propa-
gated NPCs
Heatmap and gene clustering of NPCs under
the conditions listed below. Human NPCs
from a 16- week embryonic kidney were used
as a positive control (Lindstrom et al.,
2018b). I+/P/SIX2+, ITGA8+/PDGFRA/
SIX2-GFP+ cells before expansion; I+/P/
SIX2, ITGA8+/PDGFRA/SIX2-GFP cells
before expansion; h16wk SIX2+/MEIS1+,
hMARIS_SIX2+_MEIS1+ cells from a human
16-week embryonic kidney (Lindstrom et al.,
2018b); h16wk SIX2+, hMARIS_SIX2+ cells
from a human 16-week embryonic kidney
(Lindstrom et al., 2018b); h16wk ITGA8+,
ITGA8+ cells from a human 17-week embry-
onic kidney (O’Brien et al., 2016); ActivinA
Day 7, ITGA8+/PDGFRA/SIX2-GFP+ cells
cultured with activin for 7 days; BMP7 Day 7,
ITGA8+/PDGFRA/SIX2-GFP+ cells cultured
with BMP7 for 7 days. NPC markers (Park
et al., 2012) are shown in red. Human-spe-
cific NPC markers (Lindstrom et al., 2018b)
are shown in blue.
See also Table S1.

(Figures 5A–5C). Depletion of DAPT significantly reduced findings in mouse progenitor culture (Tanigawa et al.,
the percentage of the progenitor fraction, as well as the 2016). Elimination of LIF or CHIR did not affect the per-
numbers of progenitor cells. FGF9 elimination resulted in centage of the progenitor fraction (Figures 5A and 5B),
a sharp drop in the total cell numbers, indicating that but the numbers of progenitor cells were significantly
this factor is essential for the proliferation of both progen- decreased (Figure 5C). Thus, each factor is necessary for
itors and non-progenitors, consistent with our previous proper NPC maintenance and propagation.

328 Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019


Figure 5. Activin-Mediated Signal Is Essential for Maintenance of NPCs
(A) Quantification of ITGA8+/PDGFRA/SIX2-GFP+ (I+/P/SIX2+) cells after culture for 7 days. NPCs were cultured for 7 days in the
optimized medium including activin (50 ng/mL) as indicated (All), and with individual factors eliminated from the optimized medium,
then analyzed by flow cytometry. The percentages of ITGA8+/PDGFRA/SIX2-GFP+ cells in the total cells are shown on the right (I+/P/
SIX2+ cells/total cells) (n = 3 per group). *p < 0.05, **p < 0.01 versus All.
(B) Percentages of ITGA8+/PDGFRA/SIX2-GFP+ cells among the total cells (n = 3 per group). **p < 0.01 versus All.
(legend continued on next page)

Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019 329


When the expanded cells at day 7 were passaged and of SMAD2/3, but not SMAD1/5, is correlated with NPC
cultured for a further 7 days (day 14), NPCs increased by maintenance in our protocol.
25-fold and constituted 79.4% of the expanded cells (Fig- It was reported that non-canonical BMP signaling mole-
ure 5D). Furthermore, progenitor cells retained comparable cules, namely JNK and p38, play critical roles in the main-
nephron-forming potential to day 7 (Figures S5A–S5C). tenance and proliferation of NPCs (Brown et al., 2011; Mu-
However, the percentage of the ITGA8+/PDGFRA/SIX2- thukrishnan et al., 2015). However, the phosphorylation of
GFP+ fraction at day 21 (third generation of culture) declined p38 and JNK was unaltered by LDN193189 or A83-01 treat-
to 57.3%, although the SIX2-GFP+ fraction remained high ment (Figure 5E). In addition, p38 or JNK inhibitors had
(91.5%; Figure S5D), indicating that further fine-tuning or minimal impacts on the NPC percentages (Figure 5G), indi-
addition of an unknown signal is necessary for infinite cating that non-canonical BMP signaling is not signifi-
maintenance of nephron progenitors. We also attempted cantly involved in our activin-based NPC maintenance.
monolayer cultures of single cells, but the percentage of Thus, the canonical SMAD2/3 pathway downstream of ac-
the ITGA8+/PDGFRA/SIX2-GFP+ fraction at day 14 (second tivin may play a major role in the maintenance of human
generation of culture) declined to 37.4% (Figures S5E and iPSC-derived NPCs.
S5F), indicating that three-dimensional aggregation culture
was better than two-dimensional culture. Propagated NPCs Form Large Areas of Nephrons with
Vascularized Glomeruli upon Transplantation
SMAD2/3 Phosphorylation Is Correlated with NPC Next, we examined the in vivo nephron-forming potential
Maintenance of the expanded NPCs. We cultured NPCs with activin for
To elucidate the mechanisms underlying the maintenance 7 days, combined the cells with spinal cord for a further
of NPCs, we examined the effects of BMP or TGFb recep- 2 days to initiate nephrogenesis, and transplanted the
tor inhibitors. When NPCs were cultured for 7 days using spheres underneath the kidney capsule of immunodefi-
the activin-based protocol in the presence of BMP recep- cient mice, as previously described (Sharmin et al., 2016).
tor inhibitor LDN193189 the phosphorylation levels of In this setting, it is known that iPSC-derived NPCs can
SMAD1/5 were reduced, while the percentage of the form glomeruli and renal tubules, the former of which
ITGA8+/PDGFRA/SIX2-GFP+ fraction was not affected are vascularized by host endothelial cells. Glomerular po-
(Figures 5E and 5F), indicating that BMP receptor-medi- docytes become more mature than those in vitro, and
ated SMAD1/5 activity had a minimal effect on NPC NEPHRIN is expressed at the basal side of the podocytes
maintenance. In contrast, treatment with TGFb receptor to form slit diaphragms, comprising molecular sieves for
inhibitor A83-01 resulted in a reduction of SMAD2/3 filtration from the vasculature (Tanigawa et al., 2018). As
phosphorylation and a decrease in the percentage of the a control, we used whole spheres containing NPCs before
ITGA8+/PDGFRA/SIX2-GFP+ fraction (Figures 5E and maintenance culture, as described in our previous report
5F). Consistent with the latter observation, SIX2 expres- (Sharmin et al., 2016). At day 20 after transplantation,
sion was reduced while E-cadherin was increased (Fig- the control cells showed formation of glomeruli and renal
ure 5E). Unexpectedly, we observed increased SMAD1/5 tubules (day 0 in Figure 6A). However, these nephrons
phosphorylation upon A83-01 treatment (Figure 5E), constituted only a subset (10.3%, n = 3) of the transplanted
which may reflect the higher percentage of differentiated tissues (Figures 6A and 6B). In contrast, nephrons derived
cells in the samples, because SMAD1/5 activity is known from the expanded cells occupied larger areas of the trans-
to be detectable in differentiating nephrons (Brown planted tissues (70.1%, n = 3), suggesting good preserva-
et al., 2013; Fetting et al., 2014). Thus, phosphorylation tion of the NPC percentage and potential. In addition,

(C) Numbers of total and ITGA8+/PDGFRA/SIX2-GFP+ cells after culture for 7 days (n = 3 per group). *p < 0.05, **p < 0.01 versus All.
(D) Estimated numbers of total and ITGA8+/PDGFRA/SIX2-GFP+ cells after culture for 7 or 14 days. Data represent means ± SEM from three
independent experiments.
(E) Western blotting analysis of cultured ITGA8+/PDGFRA/SIX2-GFP+ cells after treatment with BMP or TGFb receptor inhibitors. NPCs
were cultured in activin-containing medium with or without () inhibitors at the indicated concentrations for 7 days. The indicated
molecules were detected by immunoblotting.
(F) Quantification of ITGA8+/PDGFRA/SIX2-GFP+ cells after culture for 7 days. NPCs were cultured for 7 days in activin-containing medium
with or without (control) inhibitors at the indicated concentrations. The percentages of ITGA8+/PDGFRA/SIX2-GFP+ cells were measured
by flow cytometry. Data represent means ± SEM from three independent experiments. *p < 0.05, **p < 0.01 versus control.
(G) NPCs were cultured for 7 days in activin-containing medium with p38 inhibitor SB202190 (1 mM) or JNK inhibitor SP600125 (1 mM). The
percentages of ITGA8+/PDGFRA/SIX2-GFP+ cells were measured by flow cytometry (n = 3 per group).
See also Figure S5.

330 Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019


Figure 6. Propagated NPCs Form Large
Areas of Nephrons with Vascularized
Glomeruli upon Transplantation
(A) H&E-stained sections of tissues at
20 days after transplantation. Left panels:
transplants from iPSC-derived NPCs before
expansion culture (without sorting). Right
panels: transplants from ITGA8+/PDGFRA/
SIX2-GFP+ cells cultured with activin for
7 days. Scale bars, 50 mm. The bottom panels
show magnified images of the squares in the
upper panels. Scale bars, 200 mm.
(B) Quantification of nephron areas in
induced tissues. The percentages of nephron
structures in the induced tissues relative to
the total section areas were calculated and
shown as bar graphs (n = 3 per group).
(C) Magnified image of a glomerulus in
transplants of ITGA8+/PDGFRA/SIX2-GFP+
cells. Scale bar, 50 mm.
(D) Immunostaining of glomeruli. Trans-
plants of ITGA8+/PDGFRA/SIX2-GFP+ cells
after 20 and 40 days were stained for
NEPHRIN (red) and WT1 (green) or CD31
(green). The immunostained images show
vascularized glomeruli and basolateral
localization of NEPHRIN at 20 days (left and
center) and 40 days (right) after trans-
plantation. Arrowheads indicate lateral pre-
slit diaphragm domains; arrows indicate
basal linear expression of NEPHRIN. Scale
bars, 25 mm (left); and 8 mm (center and
right).

NEPHRIN was expressed in glomerular podocytes at their Sendai virus vector. Both iPSC lines had normal karyotypes
basal side and aligned in a linear manner along the endo- and expressed pluripotent stem cell markers (Figures 7A,
thelial cells (Figures 6C and 6D), which is characteristic of 7B, and S6).
maturation-stage podocytes equipped with basally located First, we induced the RN7 line toward the kidney lineage,
slit diaphragms (Tanigawa et al., 2018). These data indicate sorted the ITGA8+/PDGFRA cells at day 13, and cultured
that the ITGA8+/PDGFRA/SIX2-GFP+ NPCs propagated by them for 7 days with activin or BMP7. In the presence of ac-
activin retain their nephron-forming competence in vivo tivin, cells were expanded by 11-fold and 90% of the cells
and can successfully generate maturation-stage glomerular were ITGA8+/PDGFRA (Figure 7C). In contrast, only a
podocytes, further supporting the authenticity of our 4.4-fold increase was observed in the presence of BMP7,
protocol. although 84% of the expanded cells were ITGA8+/
PDGFRA (Figure 7C). Intriguingly, staining of cytospun
The Activin-Based Protocol Is Applicable to iPSC Lines cells from the ITGA8+/PDGFRA fraction revealed that
without a GFP Tag 85.1% of cells were SIX2+ in the presence of activin,
Next, we examined whether our protocol was applicable to compared with only 3.4% of SIX2+ cells in the presence
multiple iPSC lines. We further investigated the expansion of BMP7 (Figure 7D). The expression level of SIX2 was
of NPCs from iPSCs without a GFP tag, which would make significantly reduced in the BMP7 condition (Figure 7E).
our protocol more useful. For this, we established two new When the cells expanded by activin were induced for
iPSC lines (RN7 and RN12) from a healthy donor using a differentiation, they readily formed glomeruli and renal

Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019 331


(legend on next page)

332 Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019


tubules, and the percentages of the nephron area were improved protocol depends on quantitative measurement
larger than those of cells cultured with BMP7 (Figure 7F, of nephron progenitors based on three markers. While
n = 6). Finally, the expanded cells were transplanted under monitoring of SIX2 expression with GFP is useful, we
the renal capsules of immunodeficient mice as described demonstrated that dependence on SIX2 expression alone
above. At day 20 after transplantation, the cells expanded is insufficient and further combination with two other
by activin showed robust formation of glomeruli and renal markers (ITGA8+/PDGFRA) is essential for assessment of
tubules (Figures 7G and 7H, n = 4). The percentages of the nephron-forming competence.
nephron area in the transplanted tissues were larger than Based on these stringent criteria, we identified that
those of cells cultured with BMP7 (Figure 7G, n = 4). Round activin is superior to BMP7 in maintaining human iPSC-
and vascularized glomeruli were formed that resembled derived NPCs. We and others previously reported the
those in transplanted human embryonic kidneys (Dekel importance of BMP7 for NPC propagation in both mice
et al., 2003). The glomeruli contained podocytes positive and humans. Li et al. (2016) claimed that human iPSC-
for PODOCALYXIN (PODXL), and NEPHRIN was linearly derived SIX2-GFP+ cells are expandable for 2 months.
aligned with CD31+ vascular endothelial cells (Figure 7I). However, when we applied the same protocol, the percent-
In contrast, cells expanded by BMP7 showed minimal for- age of ITGA8+/PDGFRA/SIX2-GFP+ cells was low, while
mation of nephrons upon transplantation (Figure 7G). All that of SIX2-GFP+ cells remained relatively high, again
of these tendencies held true for the RN12 cell line (Fig- emphasizing the importance of the progenitor criteria.
ure S6). Thus, activin exhibits superior effects to BMP7 In contrast, our activin-based protocol maintained
for NPC expansion and nephrogenesis in vitro and in vivo, 80%–90% of the expanded cells as ITGA8+/PDGFRA/
and our protocol is applicable to multiple independent SIX2-GFP+. Upon transplantation, nephron epithelia
iPSC lines as well as genetically untagged clones. derived from the expanded progenitors occupied large
areas of the induced tissues and glomerular podocytes
exhibited the characteristic features of those in vivo:
DISCUSSION NEPHRIN was localized at the basal region of the podocytes
and aligned with the podocyte-endothelial boundary,
We have demonstrated that activin exerts a superior underscoring the high percentage and quality of our
effect to BMP7 on the maintenance of NPCs derived from expanded nephron progenitors. In addition, we demon-
human iPSCs. The percentage of ITGA8+/PDGFRA/SIX2- strated that the expanded cells could be frozen without
GFP+ progenitors remained high in our activin-based pro- losing their nephron-forming potential, suggesting that
tocol, and the resulting nephrons occupied the majority the complicated induction processes for NPCs can be by-
of the induced tissues both in vitro and in vivo. Our passed for certain sets of experiments. Furthermore, our

Figure 7. The Activin-Based Protocol Is Applicable to iPSC Lines without a GFP Tag
(A) Normal karyotype of an established iPSC clone (RN7) from a healthy donor.
(B) Expression of stem cell markers in RN7 iPSCs. Scale bars, 200 mm.
(C) Flow-cytometry analysis of the ITGA8+/PDGFRA fraction after a 7- day expansion culture of NPCs induced from RN7. Day 0: ITGA8+/
PDGFRA fraction of induced NPCs before expansion culture. The percentages of the ITGA8+/PDGFRA fraction after 7 days of culture with
activin or BMP7 are shown (n = 3 per group). Rightmost panel: numbers of total and ITGA8+/PDGFRA (I+/P) cells after 7 days of culture
(n = 3 per group). **p < 0.01, cells cultured with activin versus BMP7.
(D) SIX2 staining of cytospun ITGA8+/PDGFRA cells at day 7 of expansion culture. The percentages of SIX2+ cells among the ITGA8+/
PDGFRA cells are also shown (n = 3 per group).
(E) qPCR analysis of SIX2 expression in ITGA8+/PDGFRA cells after culture for 7 days. Freshly isolated ITGA8+/PDGFRA cells were used as a
positive control (Day 0). **p < 0.01, cells cultured with activin or BMP7 versus Day 0 (n = 3 per group).
(F) Immunostaining of nephron structures generated from RN7-derived ITGA8+/PDGFRA cells expanded by activin or BMP7 for 7 days.
Scale bars: 200 mm. Right panel: percentages of nephron-like structures in the induced tissues are shown as bar graphs. Data represent
means ± SEM from three independent experiments. **p < 0.01, cells cultured with activin versus BMP7.
(G) H&E-stained sections of kidney tissues at 20 days after transplantation. Left panels: transplants from RN7-derived ITGA8+/PDGFRA
cells cultured with activin (left) or BMP7 (right) for 7 days. Scale bars, 100 mm. Rightmost panel: percentages of nephron areas in
transplanted tissues (n = 4 per group). **p < 0.01, cells cultured with activin versus BMP7.
(H) Immunostaining of nephron structures (upper panel) and H&E-staining of a glomerulus (lower panel) in the transplants from RN7-
derived ITGA8+/PDGFRA cells cultured with activin. Scale bars, 200 mm (upper panel) and 50 mm (lower panel).
(I) Immunostaining of glomeruli in kidney tissues at 20 days after transplantation of RN7-derived NPCs. CD31 (green) and PODXL (red) or
NEPHRIN (red) are shown. Arrowheads indicate background signals of red blood cells. Scale bars, 25 mm.
See also Figure S6.

Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019 333


activin-based protocol was applicable to multiple iPSC 2 weeks of culture. The NGFR+/EPCAM fraction was also
lines, proving its robustness. used for NPC isolation from human embryonic kidneys
It was an unexpected finding that activin is superior to at 9–14 weeks (Li et al., 2016). However, our RNA-seq anal-
BMP7 for NPC maintenance. Most published maintenance ysis showed that the expression of NCAM, FZD7, and NGFR
protocols are based on the importance of BMP7, especially were relatively lower than that of ITGA8 in iPSC-derived
the non-canonical pathway downstream of BMP7, while NPCs (ITGA8+/PDGFRA/SIX2-GFP+) before expansion,
SMAD1 induces NPC differentiation. For example, Li while the same genes were expressed to some extent in
et al. (2016) reported maintenance of NPCs from human SIX2+ NPCs in human embryonic kidneys at 16 weeks (Ta-
embryos utilizing BMP7 and a BMP receptor inhibitor, re- ble S1). Indeed, Li et al. (2016) reported that NGFR was not
sulting in SMAD1 inhibition. Meanwhile, expression of ac- applicable to iPSC-derived NPCs, and our preliminary trials
tivin receptor type II or phosphorylated Smad2 in the on these three markers for isolation of iPSC-derived NPCs
nephron progenitor regions was reported in mice (Rowan were also unsuccessful. These results may indicate that arti-
et al., 2018). Furthermore, deletion of GDF11, which is ex- ficially induced NPCs are not completely identical to those
pressed in nephron progenitors and the ureteric bud, led in vivo. Alternatively, iPSC-derived NPCs could represent
to kidney hypoplasia (Esquela and Lee, 2003). These re- those at 4–5 weeks of gestation when NPCs first appear
ports may be relevant to our present finding that the acti- in vivo (Lindstrom et al., 2018a). Establishment of the
vin/GDF11/TGFb-SMAD2/3 pathway is involved in NPC gene expression profiles in human embryonic NPCs at
maintenance in vitro. Alternatively, the responsiveness to earlier time points will be helpful toward the identification
exogenous signals may differ between in vivo NPCs and of more reliable surface markers for iPSC-derived NPCs.
iPSC-derived NPCs. While further investigation is required Nonetheless, our protocol will be useful for a variety of set-
to address these possibilities, our data emphasize the prac- tings in iPSC-based research. Differentiation efficiencies
tical usefulness of the activin-based protocol, considering vary from clone to clone, and can sometimes be very low.
the limited access to human embryos. Even in these situations, our protocol allows propagation
However, our protocol did not achieve long-term propa- of NPCs with high efficiency, and the cells can be subjected
gation. Consistent with this limitation, our RNA-seq to subsequent functional assays for nephrogenesis in vitro
analysis showed incomplete preservation of the gene and in vivo. We recently reported that iPSCs derived from a
expression profiles during culture (Figure 4). For example, patient with a NEPHRIN mutation showed impaired forma-
SIX1 and OSR1 were decreased, which may explain the tion of slit diaphragms in podocytes (Tanigawa et al., 2018).
limited propagation and longevity of the nephron progen- We also established a selective podocyte induction protocol
itors under our conditions. Among the human nephron from human iPSCs (Yoshimura et al., 2019). Combined with
progenitor-specific genes that were recently identified in these two methods, our expansion protocol would be readily
human embryos, ECEL1, DAPL1, COL9A2, TNFRSF19, applicable for disease modeling and drug screening using pa-
and PCDH15 were reduced (Figure 4). To improve our pro- tient-derived iPSCs. We also reported the generation of
tocol, information on the expression levels of these genes branching ureteric buds from mouse ESCs and human iPSCs
in human embryonic NPCs at the equivalent stage to in vitro (Taguchi and Nishinakamura, 2017). When mouse
iPSC-derived NPCs is needed for reference. RNA-seq data ESC-derived nephron progenitors and ureteric bud were
at 16 weeks of gestation (Lindstrom et al., 2018b), which combined with mouse embryo-derived stromal cells, we
were used for comparison in this study, are helpful, but were able to reconstitute a higher-order kidney structure
gene expression levels in nephron progenitors change in vitro. While we need to induce stromal progenitors from
significantly over time in vivo (Chen et al., 2015). Data at human iPSCs, our method for expansion of nephron pro-
an earlier time point (4–5 weeks after fertilization), when genitors will be useful for reconstructing a three-dimen-
human nephron progenitors initially emerge, would be sional human kidney in the near future.
needed for precise comparisons. Nonetheless, our data pro- In summary, we have developed an activin-based proto-
vide the essential gene set that can confer sufficient compe- col for maintenance and propagation of iPSC-derived hu-
tence to human nephron progenitors. man NPCs at high efficiency, which can serve as a solid
Although our protocol is applicable to multiple iPSC basis for regenerative medicine of the kidney.
lines without a GFP tag, identification of additional surface
markers would improve the maintenance efficiency of
EXPERIMENTAL PROCEDURES
NPCs. It was reported that NPCs could be enriched
in NCAM+/CD133/FZD7+ or NCAM+/CD133/EpCAM Establishment of SIX2-GFP iPSCs
fractions from human fetal kidneys at 15–22 weeks (Har- We established SIX2-GFP iPSCs by transfecting the targeting vec-
ari-Steinberg et al., 2013; Pode-Shakked et al., 2016, tor, along with the TALEN expression plasmids (Sakuma et al.,
2017), and that some NPC genes were maintained during 2013), into human iPSCs (201B7) (Takahashi et al., 2007). A full

334 Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019


description of the methods is provided in Supplemental Experi- 24 h. Elimination of the exogenous genes was confirmed by nested
mental Procedures. We obtained 23 heterozygous GFP-knockin PCR of the vector sequences. RN7 and RN12 cells were adapted to
clones among 91 clones, as determined by PCR, Southern blotting, the feeder-free condition using StemFit AK02N medium and plates
and sequencing of the non-GFP-containing allele. Clone #12 was coated with iMatrix-511 silk, as described previously (Nakagawa
adapted to the feeder-free condition using StemFit AK02N medium et al., 2014; Yoshimura et al., 2017). NPC induction was performed
(Takara Bio, AJ100) and plates coated with iMatrix-511 silk (Nippi, based on our previously established protocol (Taguchi et al., 2014;
#892021), as described previously (Nakagawa et al., 2014; Yoshi- Taguchi and Nishinakamura, 2017).
mura et al., 2017), and further electroporated with a plasmid ex- All animal experiments were carried out in accordance with our
pressing Cre recombinase to delete the puromycin-resistant institutional guidelines and were approved by the Ethics Commit-
cassette. The resultant colonies were picked up in duplicate and pu- tee of Kumamoto University (#A29-040). The ethics committees
romycin-sensitive clones were expanded. The absence of the involved were the Ethics Committee for Epidemiological and Gen-
cassette was verified by PCR, and clones #12-8 and #12-12 were eral Research at the Faculty of Life Science, Kumamoto University,
used for detailed analyses. We used clone #12-12 for most of the and the Ethics Committee for Human Genome and Gene Analysis
presented data, but consistent data were obtained for clone Research at the Faculty of Life Science, Kumamoto University
#12-8 (see Figure S3). (approval numbers 1453 and 359, respectively).

Propagation of Human iPSC-Derived NPCs Statistical Analysis


NPC induction was performed based on our previously established All experiments were performed independently at least three
protocol (Taguchi et al., 2014; Taguchi and Nishinakamura, 2017). times. Data are presented as mean ± SEM. Data were evaluated by
Induced NPCs were dissociated by incubation with AccuMAX Student’s t test. Differences were considered significant for values
(Millipore) for 8 min, and IGTA8+/PGDFRA/SIX2-GFP+ NPCs of p < 0.01.
were sorted as described by Kaku et al. (2017). The sorted NPCs
were aggregated as described by Tanigawa et al. (2016) in the ACCESSION NUMBERS
following maintenance medium. The maintenance medium for
NPCs comprised 200 ng/mL recombinant human FGF9 ( R&D Sys- The accession number for the RNA-seq data deposited with the Na-
tems), 10 ng/mL recombinant human TGFa (Peprotech), 1 mg/mL tional Center for Biotechnology Information Gene Expression
heparin (Sigma), 10 mM Y27632 (Wako Chemicals), 500 nM Omnibus is GEO: GSE118729.
CHIR99021 (Axon Medchem), 5 ng/mL recombinant human LIF
(Millipore), 500 nM DAPT (Millipore), and 50 ng/mL recombinant SUPPLEMENTAL INFORMATION
human activin A (R&D Systems) in the medium used for NPC
Supplemental Information can be found online at https://doi.org/
induction from iPSCs (Taguchi et al., 2014). The chemical com-
10.1016/j.stemcr.2019.07.003.
pounds investigated for NPC culture were as follows: A83-01 (Toc-
ris, #2939); LDN193189 (Wako, #126-05851); SB202190 (Sigma,
AUTHOR CONTRIBUTIONS
#S7067); and SP600125 (Wako, #198-14821). For passaging, the
cells were rinsed with PBS, dissociated with AccuMAX for 8 min, Culture and analysis of iPSC-derived NPCs, S.T.; generation of
and split at a 1:3 ratio for passage every 7 days. For cryopreserva- SIX2-GFP iPSCs, Y.K.; transplantation, H.N.; TALEN construction,
tion, 7- day cultured cells were rinsed with PBS and dissociated T.S. and T.Y.; providing NPC induction protocol, A.T.; writing,
with AccuMAX for 8 min. After centrifugation, the cells were S.T., A.T., and R.N.; Funding acquisition, S.T. and R.N.; Project
washed with medium containing 50 ng/mL DNase I, resuspended administration, R.N.
(100,000 cells/vial) in Stem Cell Banker (Nippon Zenyaku Kogyo),
and stored in liquid nitrogen after 24 h of pre-storage at 80 C. De- ACKNOWLEDGMENTS
frosted cells were incubated overnight in maintenance medium to
We thank Tomoko Ohmori, Yumi Soejima, Koichiro Miike, and
allow aggregate formation on low-binding 96-well plates (Tani-
Sayoko Fujimura for technical assistance, and Drs. Masaki Hosoya
gawa et al., 2016). NPC aggregates were cultured with embryonic
and Yoshiharu Sato for the RNA-seq analysis. We also thank Alison
spinal cord for 3 days to initiate differentiation, transferred to
Sherwin, PhD, from Edanz Group (www.edanzediting.com/ac) for
transwell plates, and cultured for an additional 9–20 days in previ-
editing a draft of the manuscript. This study was supported, in part,
ously described medium (Li et al., 2016) containing 5% KnockOut
by KAKENHI grants (JP16K19494 to S.T. and JP17H06177 to R.N.)
Serum Replacement (Thermo Fisher Scientific).
from the Japan Society for the Promotion of Science, and a grant
from the Research Center Network for Realization of Regenerative
Establishment of New iPSC Lines Medicine, Japan Agency for Medical Research and Development.
Two iPSC lines (RN7 and RN12) were established from the periph- This work was also supported, in part, by The Mochida Memorial
eral blood of a healthy volunteer using a previously described Foundation for Medical and Pharmaceutical Research.
method (Soga et al., 2015). In brief, four reprogramming transcrip-
tion factors (OCT3/4, SOX2, KLF4, and MYC) were introduced us- Received: August 17, 2018
ing a Sendai virus vector. After establishing the iPSC clones on Revised: July 1, 2019
feeder cells (murine embryonic fibroblasts), the temperature-sensi- Accepted: July 1, 2019
tive Sendai virus was eliminated by culturing the cells at 38 C for Published: August 1, 2019

Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019 335


REFERENCES Lindstrom, N.O., McMahon, J.A., Guo, J., Tran, T., Guo, Q., Rut-
ledge, E., Parvez, R.K., Saribekyan, G., Schuler, R.E., Liao, C.,
Barak, H., Huh, S.H., Chen, S., Jeanpierre, C., Martinovic, J., Pari-
et al. (2018a). Conserved and divergent features of human and
sot, M., Bole-Feysot, C., Nitschke, P., Salomon, R., Antignac, C.,
mouse kidney organogenesis. J. Am. Soc. Nephrol. 29, 785–805.
et al. (2012). FGF9 and FGF20 maintain the stemness of nephron
progenitors in mice and man. Dev. Cell 22, 1191–1207. Lindstrom, N.O., Guo, J., Kim, A.D., Tran, T., Guo, Q., De Sena
Brandine, G., Ransick, A., Parvez, R.K., Thornton, M.E., Basking,
Brown, A.C., Adams, D., de Caestecker, M., Yang, X., Friesel, R., and
L., et al. (2018b). Conserved and divergent features of mesen-
Oxburgh, L. (2011). FGF/EGF signaling regulates the renewal of
chymal progenitor cell types within the cortical nephrogenic
early nephron progenitors during embryonic development. Devel-
niche of the human and mouse kidney. J. Am. Soc. Nephrol. 29,
opment 138, 5099–5112.
806–824.
Brown, A.C., Muthukrishnan, S.D., Guay, J.A., Adams, D.C., Scha-
Morizane, R., Lam, A.Q., Freedman, B.S., Kishi, S., Valerius, M.T.,
fer, D.A., Fetting, J.L., and Oxburgh, L. (2013). Role for compart-
and Bonventre, J.V. (2015). Nephron organoids derived from hu-
mentalization in nephron progenitor differentiation. Proc. Natl.
man pluripotent stem cells model kidney development and injury.
Acad. Sci. U S A 110, 4640–4645.
Nat. Biotechnol. 33, 1193–1200.
Brown, A.C., Muthukrishnan, S.D., and Oxburgh, L. (2015). A syn-
Muller, U., Wang, D., Denda, S., Meneses, J.J., Pedersen, R.A., and
thetic niche for nephron progenitor cells. Dev. Cell 34, 229–241.
Reichardt, L.F. (1997). Integrin alpha8beta1 is critically important
Chen, S., Brunskill, E.W., Potter, S.S., Dexheimer, P.J., Salomonis, for epithelial-mesenchymal interactions during kidney morpho-
N., Aronow, B.J., Hong, C.I., Zhang, T., and Kopan, R. (2015). genesis. Cell 88, 603–613.
Intrinsic age-dependent changes and cell-cell contacts regulate
Muthukrishnan, S.D., Yang, X., Friesel, R., and Oxburgh, L. (2015).
nephron progenitor lifespan. Dev. Cell 35, 49–62.
Concurrent BMP7 and FGF9 signalling governs AP-1 function to
Costantini, F., and Kopan, R. (2010). Patterning a complex organ: promote self-renewal of nephron progenitor cells. Nat. Commun.
branching morphogenesis and nephron segmentation in kidney 6, 10027.
development. Dev. Cell 18, 698–712.
Nakagawa, M., Taniguchi, Y., Senda, S., Takizawa, N., Ichisaka, T.,
Dekel, B., Burakova, T., Arditti, F.D., Reich-Zeliger, S., Milstein, O., Asano, K., Morizane, A., Doi, D., Takahashi, J., Nishizawa, M.,
Aviel-Ronen, S., Rechavi, G., Friedman, N., Kaminski, N., Passwell, et al. (2014). A novel efficient feeder-free culture system for the
J.H., et al. (2003). Human and porcine early kidney precursors as a derivation of human induced pluripotent stem cells. Sci. Rep. 4,
new source for transplantation. Nat. Med. 9, 53–60. 3594.
Esquela, A.F., and Lee, S.J. (2003). Regulation of metanephric kid- O’Brien, L.L., Guo, Q., Lee, Y., Tran, T., Benazet, J.D., Whitney, P.H.,
ney development by growth/differentiation factor 11. Dev. Biol. Valouev, A., and McMahon, A.P. (2016). Differential regulation of
257, 356–370. mouse and human nephron progenitors by the Six family of tran-
Fetting, J.L., Guay, J.A., Karolak, M.J., Iozzo, R.V., Adams, D.C., scriptional regulators. Development 143, 595–608.
Maridas, D.E., Brown, A.C., and Oxburgh, L. (2014). FOXD1 pro- Park, J.S., Ma, W., O’Brien, L.L., Chung, E., Guo, J.J., Cheng, J.G.,
motes nephron progenitor differentiation by repressing decorin Valerius, M.T., McMahon, J.A., Wong, W.H., and McMahon, A.P.
in the embryonic kidney. Development 141, 17–27. (2012). Six2 and Wnt regulate self-renewal and commitment of
Harari-Steinberg, O., Metsuyanim, S., Omer, D., Gnatek, Y., Ger- nephron progenitors through shared gene regulatory networks.
shon, R., Pri-Chen, S., Ozdemir, D.D., Lerenthal, Y., Noiman, T., Dev. Cell 23, 637–651.
Ben-Hur, H., et al. (2013). Identification of human nephron pro- Pode-Shakked, N., Gershon, R., Tam, G., Omer, D., Gnatek, Y.,
genitors capable of generation of kidney structures and functional Kanter, I., Oriel, S., Katz, G., Harari-Steinberg, O., Kalisky, T.,
repair of chronic renal disease. EMBO Mol. Med. 5, 1556–1568. et al. (2017). Evidence of in vitro preservation of human nephro-
Hartman, H.A., Lai, H.L., and Patterson, L.T. (2007). Cessation of genesis at the single-cell level. Stem Cell Reports 9, 279–291.
renal morphogenesis in mice. Dev. Biol. 310, 379–387. Pode-Shakked, N., Pleniceanu, O., Gershon, R., Shukrun, R.,
Kaku, Y., Taguchi, A., Tanigawa, S., Haque, F., Sakuma, T., Yama- Kanter, I., Bucris, E., Pode-Shakked, B., Tam, G., Tam, H., Caspi,
moto, T., and Nishinakamura, R. (2017). PAX2 is dispensable for R., et al. (2016). Dissecting stages of human kidney development
in vitro nephron formation from human induced pluripotent and tumorigenesis with surface markers affords simple prospective
stem cells. Sci. Rep. 7, 4554. purification of nephron stem cells. Sci. Rep. 6, 23562.
Kobayashi, A., Valerius, M.T., Mugford, J.W., Carroll, T.J., Self, M., Rowan, C.J., Li, W., Martirosyan, H., Erwood, S., Hu, D., Kim, Y.K.,
Oliver, G., and McMahon, A.P. (2008). Six2 defines and regulates Sheybani-Deloui, S., Mulder, J., Blake, J., Chen, L., et al. (2018).
a multipotent self-renewing nephron progenitor population Hedgehog-GLI signaling in Foxd1-positive stromal cells promotes
throughout mammalian kidney development. Cell Stem Cell 3, murine nephrogenesis via TGFbeta signaling. Development 145.
169–181. https://doi.org/10.1242/dev.159947.
Li, Z., Araoka, T., Wu, J., Liao, H.K., Li, M., Lazo, M., Zhou, B., Sui, Sakuma, T., Ochiai, H., Kaneko, T., Mashimo, T., Tokumasu, D., Sa-
Y., Wu, M.Z., Tamura, I., et al. (2016). 3D culture supports long- kane, Y., Suzuki, K., Miyamoto, T., Sakamoto, N., Matsuura, S., et al.
term expansion of mouse and human nephrogenic progenitors. (2013). Repeating pattern of non-RVD variations in DNA-binding
Cell Stem Cell 19, 516–529. modules enhances TALEN activity. Sci. Rep. 3, 3379.

336 Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019


Sharmin, S., Taguchi, A., Kaku, Y., Yoshimura, Y., Ohmori, T., Sa- (2016). Kidney organoids from human iPS cells contain multiple
kuma, T., Mukoyama, M., Yamamoto, T., Kurihara, H., and Nishi- lineages and model human nephrogenesis. Nature 536, 238.
nakamura, R. (2016). Human induced pluripotent stem cell-
Tanigawa, S., Islam, M., Sharmin, S., Naganuma, H., Yoshimura, Y.,
derived podocytes mature into vascularized glomeruli upon exper-
Haque, F., Era, T., Nakazato, H., Nakanishi, K., Sakuma, T., et al.
imental transplantation. J. Am. Soc. Nephrol. 27, 1778–1791.
(2018). Organoids from nephrotic disease-derived iPSCs identify
Soga, M., Ishitsuka, Y., Hamasaki, M., Yoneda, K., Furuya, H., Mat- impaired NEPHRIN localization and slit diaphragm formation in
suo, M., Ihn, H., Fusaki, N., Nakamura, K., Nakagata, N., et al. kidney podocytes. Stem Cell Reports 11, 727–740.
(2015). HPGCD outperforms HPBCD as a potential treatment for
Tanigawa, S., Sharma, N., Hall, M.D., Nishinakamura, R., and Per-
Niemann-Pick disease type C during disease modeling with iPS
antoni, A.O. (2015). Preferential propagation of competent SIX2+
cells. Stem Cells 33, 1075–1088.
nephronic progenitors by LIF/ROCKi treatment of the meta-
Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., To-
nephric mesenchyme. Stem Cell Reports 5, 435–447.
moda, K., and Yamanaka, S. (2007). Induction of pluripotent stem
cells from adult human fibroblasts by defined factors. Cell 131, Tanigawa, S., Taguchi, A., Sharma, N., Perantoni, A.O., and Nishi-
861–872. nakamura, R. (2016). Selective in vitro propagation of nephron
progenitors derived from embryos and pluripotent stem cells.
Taguchi, A., Kaku, Y., Ohmori, T., Sharmin, S., Ogawa, M., Sasaki,
Cell Rep. 15, 801–813.
H., and Nishinakamura, R. (2014). Redefining the in vivo origin
of metanephric nephron progenitors enables generation of com- Yoshimura, Y., Taguchi, A., and Nishinakamura, R. (2017). Gener-
plex kidney structures from pluripotent stem cells. Cell Stem Cell ation of a three-dimensional kidney structure from pluripotent
14, 53–67. stem cells. Methods Mol. Biol. 1597, 179–193.
Taguchi, A., and Nishinakamura, R. (2017). Higher-order kidney Yoshimura, Y., Taguchi, A., Tanigawa, S., Yatsuda, J., Kamba, T., Ta-
organogenesis from pluripotent stem cells. Cell Stem Cell 21, kahashi, S., Kurihara, H., Mukoyama, M., and Nishinakamura, R.
730–746. (2019). Manipulation of nephron-patterning signals enables selec-
Takasato, M., Er, P.X., Chiu, H.S., Maier, B., Baillie, G.J., Ferguson, tive induction of podocytes from human pluripotent stem cells.
C., Parton, R.G., Wolvetang, E.J., Roost, M.S., Lopes, S.M., et al. J. Am. Soc. Nephrol. 30, 304–321.

Stem Cell Reports j Vol. 13 j 322–337 j August 13, 2019 337

You might also like